WO1995013701A1 - Novel fas protein and methods of use thereof - Google Patents

Novel fas protein and methods of use thereof Download PDF

Info

Publication number
WO1995013701A1
WO1995013701A1 PCT/US1994/013173 US9413173W WO9513701A1 WO 1995013701 A1 WO1995013701 A1 WO 1995013701A1 US 9413173 W US9413173 W US 9413173W WO 9513701 A1 WO9513701 A1 WO 9513701A1
Authority
WO
WIPO (PCT)
Prior art keywords
fasδtm
fas
protein
biological sample
cell
Prior art date
Application number
PCT/US1994/013173
Other languages
French (fr)
Inventor
Philip J. Barr
John P. Shapiro
Michael C. Kiefer
Original Assignee
Lxr Biotechnology Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22542947&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1995013701(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Lxr Biotechnology Inc. filed Critical Lxr Biotechnology Inc.
Priority to JP7514564A priority Critical patent/JPH09510864A/en
Priority to AU10569/95A priority patent/AU1056995A/en
Priority to EP95901253A priority patent/EP0729300A4/en
Priority to KR1019960702558A priority patent/KR960705929A/en
Publication of WO1995013701A1 publication Critical patent/WO1995013701A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus

Definitions

  • This invention relates generally to the field of apoptosis and specifically to a novel Fas protein.
  • Apoptosis is a normal physiologic process that determines individual cell death and ultimate deletion of the cell from tissue. For review see, Apoptosis the Molecular Basis of Cell Death. Tomei and Cope, eds., Current Communications in Cell and Molecular Biology 3, Cold Spring Harbor Laboratory Press, NY, 1991. Apoptosis is a process of programmed cell death involved in a variety of normal and pathogenic biological events and can be induced by a number of unrelated stimuli. Changes in the biological regulation of apoptosis also occur during aging and are responsible for many of the conditions and diseases related to aging.
  • Fas also known as APO-1, is a cell surface protein belonging to the tumor necrosis factor/nerve growth factor receptor family, each of whose members have been shown to be capable of mediating apoptosis.
  • the cloning of Fas is described in PCT publication No. WO 91/10448; and European Patent Application Publication Number 0510691.
  • Fas is a transmembrane (TM) protein of 34,971 deduced molecular weight and an apparent molecular weight of about 45,000 which may be due to glycosylation.
  • the mature Fas molecule consists of 319 amino acid residues of which 157 are extracellular, 17 constitute the TM domain and 145 are intracellular.
  • a variety of cell types express Fas on their surface. Interestingly, Fas expression is increased in activated T-cells including CD4 + and CD8 + cells.
  • T cell surface Fas have also been associated with tumor cells and HIV-infected cells. HIV-infected cells are more sensitive to anti-Fas antibodies, yet the significance of the association of Fas with HIV infection has not yet been determined.
  • Fas ⁇ TM A novel native form of the Fas protein, (hereinafter, Fas ⁇ TM) lacking the transmembrane region is provided.
  • DNA encoding Fas ⁇ TM and recombinant cells expressing the DNA are also provided. Diagnostic and therapeutic methods utilizing Fas ⁇ TM are also provided.
  • FIG. 1 is a schematic diagram depicting the Fas genomic DNA structure around the transmembrane (TM) region.
  • Figure 2 is a schematic diagram depicting alternate splicing of the Fas RNA to produce Fas and Fas ⁇ TM mRNA.
  • Figure 3 depicts the nucleotide and amino acid residue seguences of Fas ⁇ TM.
  • Figure 4 depicts synthetic peptides used to raise antibodies useful in the detection of biologically important Fas molecules.
  • Figure 5 depicts survival of various cell lines transformed with vectors expressing Fas ⁇ TM or control, untransformed cell lines on exposure to Fas antibodies.
  • Figure 6 depicts survival of various cell lines transformed with vectors expressing Fas ⁇ TM or control, untransformed cell lines on exposure to Fas antibodies.
  • Figure 7 depicts the amount of Fas on the surface of various cell lines transformed with vectors expressing Fas ⁇ TM or control, untransformed cell lines.
  • Figure 8 depicts the activity of recombinant Fas ⁇ TM in preventing anti-Fas antibody induced cell death.
  • the present invention is to an isolated novel, secreted form of Fas protein, hereinafter designated Fas ⁇ TM and methods of use of Fas ⁇ TM.
  • the invention further includes the cloned DNA encoding Fas ⁇ TM and recombinant cells expressing the DNA.
  • the nucleotide and amino acid residue sequences of Fas ⁇ TM are shown in Figure 3.
  • Fas ⁇ TM The geno ic structure of the Fas gene, in the appropriate region, is depicted in Figure 1.
  • the locations of the introns and exons are related to the different regions of Fas.
  • the cloning of Fas ⁇ TM is described in detail in the Examples below.
  • Figure 2 depicts the alternate splicing of the mRNA thought to result in the alternate forms of Fas however, the invention is not limited by the mechanism by which
  • Fas ⁇ TM is produced. Native Fas ⁇ TM lacks twenty-one amino acid residues including the TM region.
  • the invention includes other recombinant variations of Fas ⁇ TM which lack a portion of the TM region sufficient to produce non-membrane bound protein.
  • the protein is secreted from the cell.
  • Fas ⁇ TM encompasses all the non-membrane-bound forms of the molecule lacking TM region amino acid residues.
  • Figure 3 depicts the nucleotide and amino acid residue sequences of native Fas ⁇ TM.
  • the DNA encoding Fas ⁇ TM includes, but is not limited to, the cDNA, genome-derived DNA and synthetic or semi-synthetic DNA.
  • the nucleotide sequence of the cDNA encoding Fas ⁇ TM is shown in Figure 3.
  • the DNA includes modifications such as deletions, substitutions and additions, particularly in the noncoding regions. Such changes are useful to facilitate cloning and modify gene expression.
  • Various substitutions can be made within the coding region that either do not alter the amino acid residues encoded or result in conservatively substituted amino acid residues.
  • Nucleotide substitutions that do not alter the amino acid residues encoded are useful for optimizing gene expression in different systems. Suitable substitutions are known to those of skill in the art and are made, for instance, to reflect preferred codon usage in the particular expression systems. Suitable conservative amino acid residue substitutions are known in the art and are discussed below.
  • the invention further embodies a variety of DNA vectors having cloned therein the nucleotide sequence encoding Fas ⁇ TM.
  • Suitable vectors include any known in the art including, but not limited to, those for use in bacterial, mammalian and insect expression systems. Specific vectors are known in the art and need not be described in detail herein.
  • the vectors may also provide inducible promoters for expression of Fas ⁇ TM. Inducible promoters are those which do not allow constitutive expression of the gene but rather, permit expression only under certain circumstances. Such promoters may be induced by a variety of stimuli including, but not limited to, exposure of a cell containing the vector to a ligand, chemical or change in temperature.
  • promoters may also be cell-specific, that is, inducible only in a particular cell type and often only during a specific period of time.
  • the promoter may further be cell cycle specific, that is, induced or inducible only during a particular stage in the cell cycle.
  • the promoter may be both cell type specific and cell cycle specific. Any inducible promoter known in the art is suitable for use in the present invention.
  • the invention further includes a variety of expression systems transfected with the vectors. Suitable expression systems include, but are not limited to, bacterial, mammalian and insect. Specific expression systems are known in the art and need not be described in detail herein. It has been found that the baculovirus expression system described below provides expression of biologically active Fas ⁇ TM.
  • mammalian expression systems such as Chinese hamster ovary (CHO) cells
  • the invention encompasses cells removed from animals, including man, transfected with vectors encoding Fas ⁇ TM and reintroduced into the animal.
  • Suitable transfected cells also include those removed from a patient, transfected and reintroduced into the patient. Any cells can be treated in this manner.
  • Suitable cells include, but are not limited to, cardiomyocytes and lymphocytes. For instance, lymphocytes, removed, transfected with the recombinant DNA and reintroduced into an HIV-positive patient may increase the half-life of the reintroduced T cells.
  • the white blood cells are removed and sorted to yield the CD4 + cells.
  • the CD4 + cells are then transfected with a vector encoding Fas ⁇ TM and reintroduced into the patient.
  • the unsorted lymphocytes can be transfected with a recombinant vector encoding the Fas ⁇ TM gene under the control of a cell-specific promoter such that only CD4 + cells express the Fas ⁇ TM gene.
  • an ideal promoter would be the CD4 promoter, however, any suitable CD4 + T cell-specific promoter can be used.
  • the invention encompasses cells transfected in vivo by the vectors.
  • Suitable methods of jln vivo transfection are known in the art and include, but are not limited to, that described by Zhu et al. (1993) Science, 261:209-211.
  • the transfection is cell-type specific or that the promoter is cell-specific.
  • Transgenic animals containing the recombinant DNA vectors are also encompassed by the invention. Methods of making transgenic animals are known in the art and need not be described in detail herein. For a review of methods used to make transgenic animals, see PCT publication no. WO 93/04169. Preferably, such animals express a recombinant Fas ⁇ TM gene under control of a cell-specific and, even more preferably, a cell cycle specific promoter.
  • Fas ⁇ TM expressed either by the recombinant DNA or from biological sources such as sera can be accomplished by any method known in the art. Since native and most recombinant Fas ⁇ TM are secreted from the cells, purification is simplified by the fact that it appears in the supernatant of in vitro cultures and in sera in vivo and no cell disruption is required as is the case with Fas. Protein purification methods are known in the art. Generally, substantially purified proteins are those which are free of other, contaminating cellular substances, particularly proteins.
  • Fas ⁇ TM is more than eighty percent pure and most preferably Fas ⁇ TM is more than ninety-five percent pure. For clinical use as described below, Fas ⁇ TM is preferably highly purified, at least about ninety-nine percent pure, free of pyrogens and other contaminants.
  • Suitable methods of purification include, but are not limited to, affinity chromatography, immunoaffinity chromatography, size exclusion chromatography, HPLC and FPLC. Any purification scheme that does not result in substantial degradation of the protein is suitable for use in the present invention.
  • the invention also includes the substantially purified Fas ⁇ TM protein having the amino acid residue sequence depicted in Figure 3 and any protein lacking a sufficient portion of the TM region to be secreted from the cell.
  • the invention encompasses functionally equivalent variants of Fas ⁇ TM which do not significantly affect its properties. For instance, conservative substitutions of amino acid residues, one or a few amino acid deletions or additions, and substitution of amino acid residues by amino acid analogs are within the scope of the invention.
  • Amino acid residues which can be conservatively substituted for one another include but are not limited to: glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; serine/threonine; lysine/arginine; and phenylalanine/tyrosine. Any conservative amino acid substitution which does not significantly affect the properties of Fas ⁇ TM is encompassed by the present invention. mRNA encoding Fas ⁇ TM has been detected in a variety of human organs and tissues.
  • Fas ⁇ TM has also been found to be secreted from the cell rather than remain membrane-bound, even though it retains the cytoplasmic region of the membrane-bound form of the protein. Fas ⁇ TM can thus be detected in sera as a soluble protein. Any antibody that recognizes Fas is suitable for use in recognizing Fas ⁇ TM. In another embodiment, diagnostic methods are provided to detect the expression of Fas ⁇ TM either at the protein level or the mRNA level.
  • Fas ⁇ TM The soluble Fas ⁇ TM protein is likely to be found in the sera of patients with diseases associated with apoptosis defects, and is therefore useful as a diagnostic tool for detecting and monitoring biological conditions associated with such apoptosis defects.
  • Fas ⁇ TM can be detected by any antibody, either polyclonal or monoclonal, that recognizes Fas.
  • the distinction between Fas and Fas ⁇ TM can be determined by the solubility of the protein. Fas is membrane bound and can thus be removed from soluble proteins by removal of cells and/or membranes. Fas ⁇ TM remains soluble.
  • antibodies specific for Fas ⁇ TM and not Fas are encompassed by the present invention.
  • Fas ⁇ TM as the antigen or, preferably, peptides encompassing the region in Fas ⁇ TM that differs from Fas, the TM region. Examples of such peptides are depicted in Figure 5.
  • Methods of detecting proteins using antibodies and of generating antibodies using proteins or synthetic peptides are known in the art and need not be described in detail herein. Fas ⁇ TM protein expression can also be monitored by measuring the level of mRNA encoding Fas- ⁇ TM. Any method for detecting specific mRNA species is suitable for use in this method. This is easily accomplished using the polymerase chain reaction (PCR) .
  • PCR polymerase chain reaction
  • the primers chosen for PCR flank the TM region so as to provide a product that is measurably distinct in size between Fas and Fas ⁇ TM.
  • Northern blots can be utilized to detect the specific mRNA species either by size or by probes specific to the mRNA encoding the TM region.
  • the invention also encompasses therapeutic methods and compositions involving treatment with Fas ⁇ TM.
  • Fas ⁇ TM Either native or recombinant Fas ⁇ TM is suitable for use in this composition. Both should be substantially pure and free of pyrogens. It is preferred that the recombinant Fas ⁇ TM be produced in a mammalian cell line so as to ensure proper glycosylation. Fas ⁇ TM may also be produced in an insect cell line and will be glycosylated.
  • a therapeutically effective amount of substantially pure Fas ⁇ TM is suspended in a physiologically accepted buffer including, but not limited to, saline and phosphate buffered saline (PBS) and administered to the patient.
  • PBS phosphate buffered saline
  • administration is intravenous.
  • Other methods of administration include but are not limited to, subcutaneous, intraperitoneal, gastrointestinal and directly to a specific organ, such as intracardiac, for instance, to treat cell death related to myocardial infarction.
  • Fas ⁇ TM Suitable buffers and methods of administration are known in the art.
  • the effective concentration of Fas ⁇ TM will need to be determined empirically and will depend on the type and severity of the disease, disease progression and health of the patient. Such determinations are within the skill of one in the art.
  • Fas ⁇ TM is a human protein normally found in the sera; administration of exogenous human Fas ⁇ TM is not likely to induce reactions such as anaphylactic shock or the production of antibodies.
  • the upper concentration of Fas ⁇ TM for therapeutic use is thus not limited by these physiological considerations.
  • Fas ⁇ TM results in an increased extracellular concentration of Fas ⁇ TM which competitively binds the Fas ligand and therefore prevents or ameliorates apoptotic signals transmitted by Fas to the cell.
  • the therapeutic method thus includes, but is not limited to, inhibiting Fas-mediated cell death.
  • Fas-mediated cell death For instance, tumor necrosis factor (TNF) and Fas-specific antibodies are known to induce apoptosis and even whole animal death by binding to Fas. Inhibition of this interaction of Fas and ligands which induce it to trigger apoptosis thus will reduce apoptosis.
  • TNF tumor necrosis factor
  • Fas-specific antibodies are known to induce apoptosis and even whole animal death by binding to Fas. Inhibition of this interaction of Fas and ligands which induce it to trigger apoptosis thus will reduce apoptosis.
  • Suitable indications for therapeutic use of Fas ⁇ TM are those involving Fas-mediated cell death and include, but are not limited to, conditions in which there is inappropriate expression or up-regulation of Fas or the Fas ligand.
  • Such indications include, but are not limited to, HIV infection, autoimmune diseases, cardiomyopathies, neuronal disorders, hepatitis and other liver diseases, osteoporosis, and shock syndromes, including, but not limited to, septicemia.
  • Methods of treatment with Fas ⁇ TM also include other mechanisms of increasing the extracellular concentration of Fas ⁇ TM. These include, but are not limited to, increasing cellular expression of Fas ⁇ TM. Suitable methods of increasing cellular expression of Fas ⁇ TM include, but are not limited to, increasing endogenous expression and transfecting the cells with vectors encoding Fas ⁇ TM. Cellular transfection is discussed above and is known in the art. Increasing endogenous expression of Fas ⁇ TM can be accomplished by exposing the cells to biological modifiers that directly or indirectly increase levels of Fas ⁇ TM either by increasing expression or differential processing of the Fas ⁇ TM over Fas or by decreasing Fas ⁇ TM degradation.
  • Suitable biological modifiers can be determined by exposing cells expressing Fas ⁇ TM under the control of the native Fas promoter to potential biological modifiers and monitoring expression of Fas ⁇ TM. Expression of Fas ⁇ TM can be monitored as described above either by protein or mRNA levels.
  • Biological modifiers can be any therapeutic agent or chemical known in the art. Preferably, suitable biological modifiers are those lacking substantial cytotoxicity and carcinogenicity.
  • Fas ⁇ TM biological modifiers which reduce endogenous levels of Fas ⁇ TM are encompassed by the invention as is a method of increasing Fas-mediated cell death by decreasing endogenous levels of Fas ⁇ TM.
  • the method of determining suitable biological modifiers is as discussed above, except that the endpoint is decreased levels of Fas ⁇ TM.
  • Other methods of decreasing endogenous levels of Fas ⁇ TM include, but are not limited to, antisense nucleotide therapy and exposure to anti-FAS ⁇ TM antibody. Both these methods are known in the art and their application will be apparent to one of skill in the art.
  • Suitable indications for decreasing endogenous levels of Fas ⁇ TM will be any which Fas-mediated cell death is appropriate. These include, but are not limited to, various types of malignancies and other disorders resulting in uncontrolled cell growth such as eczema. The following examples are provided to illustrate but not limit the present invention.
  • Fas- ⁇ TM Nucleic acid sequences encoding Fas ⁇ TM were cloned as follows. Unless otherwise specified, all cloning techniques were essentially as described by Sambrook et al. (1989) and all reagents were used according to the manufacturer 's instructions. mRNA was obtained from human lymphocytes and PCR was used to make cDNA specific for the Fas ⁇ TM mRNA. The lymphocytes were obtained and processed as follows. 35 ml of blood was obtained by venipuncture from a normal 42 year old male and immediately added to 350 ⁇ l 15% EDTA.
  • RNA isolation was then processed for RNA isolation as follows. Total RNA was isolated from 3 x 10 6 cells using the guanidinium thiocyanate single-step RNA isolation method according to "Current Protocols in Molecular Biology" (1991) .
  • the first strand cDNA used in the PCR reaction was synthesized from 6.4 ⁇ g of total RNA and resuspended in 100 ⁇ l water, according to the method described by Zapf et al. (1990) J. Biol. Chem. , 265:14892-14898.
  • the Fas ⁇ TM cDNA was synthesized using PCR.
  • the forward primer: 5'-GATTGCTTCTAGACCATGCTGGGCATCTGGACCCTCCTACC-3' contained an Xbal restriction site, and encoded the initiation methionine codon and first eight codons of Fas.
  • 5'-GTTGTTTGTCGACCTAGACCAAGCTTTGGATTTCATTTCTG-3' contained a Sail restriction site, and encoded the termination codon and last eight codons of Fas.
  • the PCR reaction was performed by adding to 1 ⁇ l of template cDNA (as described above, diluted 1:100), 100 pmoles of each primer, 2.5 units Amplitaq, 76.5 ⁇ l H 2 0 and 10 ⁇ l buffer. The reaction proceeded at 94°C for 1 min, 55°C for 2 min, 72°C for 3 min for 35 cycles, 72°C for 7 min and was stored at 4°C. 15 ⁇ l of the reaction mix was loaded on a 1% agarose gel and a band of the appropriate molecular weight was detected. The remaining reaction mix was extracted with phenol/chloroform, ethanol precipitated and resuspended in 80 ⁇ l Tris-EDTA (TE) .
  • TE Tris-EDTA
  • the purified Fas ⁇ TM cDNA was resuspended in 20 ⁇ l TE buffer and the pBluescript in 40 ⁇ l TE buffer.
  • the DNA samples were ligated in a reaction mixture containing 2 ⁇ l vector, 8 ⁇ l Fas ⁇ TM cDNA, 2 ⁇ l 10 mM ATP,
  • the intron-exon organization in the Fas TM region was determined by PCR. Primers were designed to flank each of the putative introns, 1 and 2 (see Figure 2) .
  • the forward and reverse primers flanking intron 1 were 5'-GATTGCTTCTAGAGGAATCA TCAAGGAATGCACACTC-3' and 5'-GTTGTTTGTCGACC CAAACAATTAGTGGAATTGGCAA-3' respectively, and the forward and reverse primers for intron 2 were 5'-AGATCTGCGGCCGCAT TGGGGTGGCTTTGTCTTCTTCTT-3' and 5'-GTTGTTTGTCGACGTTTTCCT TTCTGTGCTTTCTGCA -3' respectively.
  • Xbal and Sail restriction enzyme sites and Notl and Sail restriction enzyme sites were included at the 5' ends of the intron 1 and 2 primers respectively to facilitate cloning of the PCR products.
  • PCR was performed according to the manufacturer's instructions (Perkin Elmer Cetus) using human genomic DNA (Clontech) (5 ⁇ g) as template. 30 cycles of PCR were performed in a Perkin Elmer Cetus DNA Thermal Cycler with each cycle consisting of a 94°C, 1 min denaturation step, a 55°C, 2 min annealing step, and a 72°C, 3 min extension step. An additional 7 minute extension step was included after the last cycle.
  • PCR products were then incubated with 5 units of DNA poly erase I, Klenow fragment at 37°C, 30 min, extracted with phenol/chloroform/isoamylalcohol (1:1:0.04) followed by chloroform/isoamylalcohol (24:1) and recovered by ethanol precipitation.
  • intron 1 and intron 2 PCR products were digested with Xbal and Sail and Notl and Sail respectively, agarose gel purified, ligated into pBluescript SK(-) and introduced into E. coli strain HB101 by the methods described in Example 1.
  • Plasmid DNA was isolated using a Promega Magic miniprep kit according to the method described by the manufacturer. Plasmid DNA was sequenced directly by the dideoxy chain termination method using the Sequenase Version 2.0 DNA Sequencing kit according to the manufacturer's instructions (USB) . The sequence obtained is depicted in Figure 3.
  • Example 3 Expression of Recombinant Fas- ⁇ TM
  • the plasmid obtained in Example 1 was used to generate a second Fas ⁇ TM vector, designated pBlueBacIII-Fas ⁇ TM, by a PCR methodology as described in Example 1.
  • the forward primer, 5'-TTTCCCGGATCCACAACCATGCTGGGCATCTGGACCCTCCTA-3' contained the convenient BamHI restriction site, and a Kozak consensus sequence ACAACC immediately preceding the initiation codon, and encoded the first seven amino acids of Fas.
  • the reverse primer 5'-CCCCATGGCTAGACCAAGCTTTG GATTTCATT-3' encoded a termination codon, a Ncol site, and the last seven amino acids of Fas.
  • Five recombinant plasmids were isolated. Two of them were sequenced by the dideoxy terminator method (Sanger et al. 1977) using sequencing kits according to the manufacturer's instructions (USB, Sequenase version 2.0). The DNA was sequenced using internal primers.
  • PAGE of the protein produced in the baculovirus system is identical to the predicted molecular mass of Fas ⁇ TM according to the amino-acid sequence and the recombinant protein was also recognized by an anti-Fas antibody (Medical and Biological Laboratories, Nagoya, Japan) .
  • Fas ⁇ TM in Mammalian Systems
  • the Fas ⁇ TM coding sequence was excised from pBluescript-Fas ⁇ TM with Xbal and Sail, and introduced into plasmids pCEP7 and pREP7 (Invitrogen) at compatible Nhel and Xhol sites to generate clones Fas ⁇ TM-1 and Fas ⁇ TM-7, respectively.
  • pCEP7 was generated by removing the RSV 3'-LTR of pREP7 with Xbal/Asp718. and substituting the CMV promoter from pCEP4 (Invitrogen) .
  • pCEP4 was first cleaved with Sail, and ligated to an oligonucleotide adapter containing an external Sail site, and an internal Nhel site. pCEP4 was then cleaved with Nhel and Asp.718 and the purified CMV promoter was ligated into pREP7 to generate pCEP7. 25 ⁇ g of each Fas ⁇ TM-containing plasmid was electroporated into the B lymphoblastoid cell line
  • WIL-2 WIL-2
  • stable hygromycin resistant transformants were selected.
  • Fas ⁇ TM-1 and Fas ⁇ TM-7 transformed WIL-2 cells were grown in RPMI supplemented with 10% fetal bovine serum (FBS) . After washing with fresh medium, the cells were suspended in RPMI supplemented with 10% FBS, 50 ng anti-Fas antibody was added, and the kinetics of cell death were analyzed by flow cytometry with FACScan. This method is based on the measurement of cells which shrink and are permeable to propidium iodide (PI) following their death. There was no difference in survival of all three cell lines in the control, but upon addition of anti-Fas antibodies, cells transformed by Fas ⁇ TM were less sensitive: by 26 hrs treatment approximately 50%, 40%, and 16% of wild type WIL-2,
  • Example 6 Recombinant Fas ⁇ TM Prevents Cell Death Induced by Anti-Fas Antibodies
  • insect cells transfected with wild type baculovirus and recombinant containing Fas ⁇ TM, as described in Example 3 were homogenized in water or in buffer containing 0.05% nonionic detergent Triton X-100 and centrifuged.
  • Anti-Fas antibodies were preincubated 2 hrs at room temperature with aliquots of soluble and insoluble fractions, added to WIL-2, and cell death was analyzed after 24 hrs by flow cytometry as described above.
  • Example 7 Fas ⁇ TM transcript analysis by RT-PCR Native Fas ⁇ TM transcripts were identified by RT-PCR and acrylamide gel electrophoresis. PCR primers were designed around the Fas TM region so that the Fas and Fas ⁇ TM transcripts would yield 296 bp and 233 bp PCR products, respectively. The forward primer was 5'-GACCCAGAATACCAAGTGCAGATGTA-3' and the reverse primer was 5'-CTGTTTCAGGATTTAAGGTTGGAGATT-3' . cDNA was synthesized from poly(A)+ or total RNA isolated from various human tissues and cell lines. These included heart, liver, activated and non-activated peripheral blood lymphocytes (PBLs) , placenta and fibroblast cell lines. PCR was performed as described in Example 1 using the cDNA as templates (10-100 ng/ml) and products were analyzed on 7% acrylamide/TBE gels.
  • PBLs peripheral blood lymphocytes

Abstract

The present invention provides a novel form of the Fas protein, DNA encoding the protein cells expressing the recombinant DNA and methods of use thereof.

Description

NOVEL FAS PROTEIN AND METHODS OF USE THEREOF
Field of the Invention
This invention relates generally to the field of apoptosis and specifically to a novel Fas protein.
Background of the Invention
Apoptosis is a normal physiologic process that determines individual cell death and ultimate deletion of the cell from tissue. For review see, Apoptosis the Molecular Basis of Cell Death. Tomei and Cope, eds., Current Communications in Cell and Molecular Biology 3, Cold Spring Harbor Laboratory Press, NY, 1991. Apoptosis is a process of programmed cell death involved in a variety of normal and pathogenic biological events and can be induced by a number of unrelated stimuli. Changes in the biological regulation of apoptosis also occur during aging and are responsible for many of the conditions and diseases related to aging.
Recent studies of apoptosis have implied that a common metabolic pathway leading to cell death may be initiated by a wide variety of signals, including changes in hormone levels, serum growth factor deprivation, chemotherapeutic agents and ionizing radiation. Wyllie (1980) Nature, 284:555-556; Kanter et al. (1984) Biochem. Biophys. Res. Commun. , 155:324-331; and Kru an et al. (1991) J. Cell. Physiol., 148:267-273. Agents that affect the biological control of apoptosis thus have therapeutic utility in a wide variety of conditions.
Fas, also known as APO-1, is a cell surface protein belonging to the tumor necrosis factor/nerve growth factor receptor family, each of whose members have been shown to be capable of mediating apoptosis. The cloning of Fas is described in PCT publication No. WO 91/10448; and European Patent Application Publication Number 0510691. Fas is a transmembrane (TM) protein of 34,971 deduced molecular weight and an apparent molecular weight of about 45,000 which may be due to glycosylation. The mature Fas molecule consists of 319 amino acid residues of which 157 are extracellular, 17 constitute the TM domain and 145 are intracellular. A variety of cell types express Fas on their surface. Interestingly, Fas expression is increased in activated T-cells including CD4+ and CD8+ cells.
Certain antibodies specific for Fas have been shown to induce death of cells that express Fas on their surfaces, by an apoptotic mechanism. Early studies indicated that therapeutic uses of antibodies specific to Fas would be effective in treating a variety of diseases. Itoh et al. (1991) Cell, 66:233-243; Krammer et al. (1991) in Apoptosis: The Molecular Basis of Cell Death. (Tomei and Cope, eds.), Cold Spring Harbor Laboratory
Press, NY; Oehm et al. (1992) J. Biol. Chem. , 267:10709- 10715; and Rouvier et al. (1993) J. Exp. Med. , 177:195- 200. It has now been found that administration of anti-Fas antibodies can be lethal. Ogasawara et al. (1993) Nature, 364:806-809. It has also been found that purified Fas blocks the cytocidal effects of anti-Fas. Oehm et al. (1992) .
Increased levels of T cell surface Fas have also been associated with tumor cells and HIV-infected cells. HIV-infected cells are more sensitive to anti-Fas antibodies, yet the significance of the association of Fas with HIV infection has not yet been determined.
The endogenous Fas ligand, responsible for recognizing Fas and inducing apoptosis, has not been identified, although some AIDS patients have been shown to have increased levels of anti-Fas autoantibodies. Oehm et al. (1991) . Moreover, T cell mediated cytotoxicity has been shown to be involved in Fas- mediated apoptosis. All references cited herein both infra and supra are hereby incorporated herein by reference. Summary of the Invention
A novel native form of the Fas protein, (hereinafter, FasΔTM) lacking the transmembrane region is provided. DNA encoding FasΔTM and recombinant cells expressing the DNA are also provided. Diagnostic and therapeutic methods utilizing FasΔTM are also provided.
Brief Description of the Drawings
Figure 1 is a schematic diagram depicting the Fas genomic DNA structure around the transmembrane (TM) region.
Figure 2 is a schematic diagram depicting alternate splicing of the Fas RNA to produce Fas and FasΔTM mRNA.
Figure 3 depicts the nucleotide and amino acid residue seguences of FasΔTM. Figure 4 depicts synthetic peptides used to raise antibodies useful in the detection of biologically important Fas molecules.
Figure 5 depicts survival of various cell lines transformed with vectors expressing FasΔTM or control, untransformed cell lines on exposure to Fas antibodies.
Figure 6 depicts survival of various cell lines transformed with vectors expressing FasΔTM or control, untransformed cell lines on exposure to Fas antibodies.
Figure 7 depicts the amount of Fas on the surface of various cell lines transformed with vectors expressing FasΔTM or control, untransformed cell lines.
Figure 8 depicts the activity of recombinant FasΔTM in preventing anti-Fas antibody induced cell death.
Detailed Description of the Invention The present invention is to an isolated novel, secreted form of Fas protein, hereinafter designated FasΔTM and methods of use of FasΔTM. The invention further includes the cloned DNA encoding FasΔTM and recombinant cells expressing the DNA. The nucleotide and amino acid residue sequences of FasΔTM are shown in Figure 3.
The geno ic structure of the Fas gene, in the appropriate region, is depicted in Figure 1. The locations of the introns and exons are related to the different regions of Fas. The cloning of FasΔTM is described in detail in the Examples below.
Figure 2 depicts the alternate splicing of the mRNA thought to result in the alternate forms of Fas however, the invention is not limited by the mechanism by which
FasΔTM is produced. Native FasΔTM lacks twenty-one amino acid residues including the TM region.
The invention includes other recombinant variations of FasΔTM which lack a portion of the TM region sufficient to produce non-membrane bound protein.
Preferably, the protein is secreted from the cell. The term FasΔTM encompasses all the non-membrane-bound forms of the molecule lacking TM region amino acid residues. Figure 3 depicts the nucleotide and amino acid residue sequences of native FasΔTM.
One embodiment of the present invention is the DNA encoding FasΔTM. The DNA encoding FasΔTM includes, but is not limited to, the cDNA, genome-derived DNA and synthetic or semi-synthetic DNA. The nucleotide sequence of the cDNA encoding FasΔTM is shown in Figure 3. The DNA includes modifications such as deletions, substitutions and additions, particularly in the noncoding regions. Such changes are useful to facilitate cloning and modify gene expression. Various substitutions can be made within the coding region that either do not alter the amino acid residues encoded or result in conservatively substituted amino acid residues. Nucleotide substitutions that do not alter the amino acid residues encoded are useful for optimizing gene expression in different systems. Suitable substitutions are known to those of skill in the art and are made, for instance, to reflect preferred codon usage in the particular expression systems. Suitable conservative amino acid residue substitutions are known in the art and are discussed below.
Techniques for nucleic acid manipulation useful for the practice of the present invention are described in a variety of references, including, but not limited to. Molecular Cloning: A Laboratory Manual. 2nd ed. , Vol. 1-3, eds. Sambrook et al. Cold Spring Harbor Laboratory Press (1989) ; and Current Protocols in Molecular Biology, eds. Ausubel et al., Greene Publishing and Wiley-Interscience: New York (1987) and periodic updates.
The invention further embodies a variety of DNA vectors having cloned therein the nucleotide sequence encoding FasΔTM. Suitable vectors include any known in the art including, but not limited to, those for use in bacterial, mammalian and insect expression systems. Specific vectors are known in the art and need not be described in detail herein. The vectors may also provide inducible promoters for expression of FasΔTM. Inducible promoters are those which do not allow constitutive expression of the gene but rather, permit expression only under certain circumstances. Such promoters may be induced by a variety of stimuli including, but not limited to, exposure of a cell containing the vector to a ligand, chemical or change in temperature.
These promoters may also be cell-specific, that is, inducible only in a particular cell type and often only during a specific period of time. The promoter may further be cell cycle specific, that is, induced or inducible only during a particular stage in the cell cycle. The promoter may be both cell type specific and cell cycle specific. Any inducible promoter known in the art is suitable for use in the present invention. The invention further includes a variety of expression systems transfected with the vectors. Suitable expression systems include, but are not limited to, bacterial, mammalian and insect. Specific expression systems are known in the art and need not be described in detail herein. It has been found that the baculovirus expression system described below provides expression of biologically active FasΔTM. For expressing FasΔTM for therapeutic purposes however, mammalian expression systems, such as Chinese hamster ovary (CHO) cells, may be preferred to ensure proper post-translational modification. The invention encompasses cells removed from animals, including man, transfected with vectors encoding FasΔTM and reintroduced into the animal. Suitable transfected cells also include those removed from a patient, transfected and reintroduced into the patient. Any cells can be treated in this manner. Suitable cells include, but are not limited to, cardiomyocytes and lymphocytes. For instance, lymphocytes, removed, transfected with the recombinant DNA and reintroduced into an HIV-positive patient may increase the half-life of the reintroduced T cells.
Preferably, for treatment of HIV-infected patients, the white blood cells are removed and sorted to yield the CD4+ cells. The CD4+ cells are then transfected with a vector encoding FasΔTM and reintroduced into the patient. Alternatively, the unsorted lymphocytes can be transfected with a recombinant vector encoding the FasΔTM gene under the control of a cell-specific promoter such that only CD4+ cells express the FasΔTM gene. In this case, an ideal promoter would be the CD4 promoter, however, any suitable CD4+ T cell-specific promoter can be used.
Further, the invention encompasses cells transfected in vivo by the vectors. Suitable methods of jln vivo transfection are known in the art and include, but are not limited to, that described by Zhu et al. (1993) Science, 261:209-211. In the case of in vivo transfection, it is preferred that the transfection is cell-type specific or that the promoter is cell-specific.
Transgenic animals containing the recombinant DNA vectors are also encompassed by the invention. Methods of making transgenic animals are known in the art and need not be described in detail herein. For a review of methods used to make transgenic animals, see PCT publication no. WO 93/04169. Preferably, such animals express a recombinant FasΔTM gene under control of a cell-specific and, even more preferably, a cell cycle specific promoter.
Purification or isolation of FasΔTM expressed either by the recombinant DNA or from biological sources such as sera can be accomplished by any method known in the art. Since native and most recombinant FasΔTM are secreted from the cells, purification is simplified by the fact that it appears in the supernatant of in vitro cultures and in sera in vivo and no cell disruption is required as is the case with Fas. Protein purification methods are known in the art. Generally, substantially purified proteins are those which are free of other, contaminating cellular substances, particularly proteins. Preferably, FasΔTM is more than eighty percent pure and most preferably FasΔTM is more than ninety-five percent pure. For clinical use as described below, FasΔTM is preferably highly purified, at least about ninety-nine percent pure, free of pyrogens and other contaminants.
Suitable methods of purification are known in the art and include, but are not limited to, affinity chromatography, immunoaffinity chromatography, size exclusion chromatography, HPLC and FPLC. Any purification scheme that does not result in substantial degradation of the protein is suitable for use in the present invention. The invention also includes the substantially purified FasΔTM protein having the amino acid residue sequence depicted in Figure 3 and any protein lacking a sufficient portion of the TM region to be secreted from the cell. The invention encompasses functionally equivalent variants of FasΔTM which do not significantly affect its properties. For instance, conservative substitutions of amino acid residues, one or a few amino acid deletions or additions, and substitution of amino acid residues by amino acid analogs are within the scope of the invention.
Amino acid residues which can be conservatively substituted for one another include but are not limited to: glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; serine/threonine; lysine/arginine; and phenylalanine/tyrosine. Any conservative amino acid substitution which does not significantly affect the properties of FasΔTM is encompassed by the present invention. mRNA encoding FasΔTM has been detected in a variety of human organs and tissues. These include liver; heart; peripheral blood lymphocytes (PBLs) , both activated and normal; placenta; fibroblasts, both normal and phorbol ester treated and SV40 infected; and several cell lines including U937, WIL-2 and IM9. FasΔTM has also been found to be secreted from the cell rather than remain membrane-bound, even though it retains the cytoplasmic region of the membrane-bound form of the protein. FasΔTM can thus be detected in sera as a soluble protein. Any antibody that recognizes Fas is suitable for use in recognizing FasΔTM. In another embodiment, diagnostic methods are provided to detect the expression of FasΔTM either at the protein level or the mRNA level. The soluble FasΔTM protein is likely to be found in the sera of patients with diseases associated with apoptosis defects, and is therefore useful as a diagnostic tool for detecting and monitoring biological conditions associated with such apoptosis defects. FasΔTM can be detected by any antibody, either polyclonal or monoclonal, that recognizes Fas. The distinction between Fas and FasΔTM can be determined by the solubility of the protein. Fas is membrane bound and can thus be removed from soluble proteins by removal of cells and/or membranes. FasΔTM remains soluble. Alternatively, antibodies specific for FasΔTM and not Fas are encompassed by the present invention. Such antibodies can be generated by using FasΔTM as the antigen or, preferably, peptides encompassing the region in FasΔTM that differs from Fas, the TM region. Examples of such peptides are depicted in Figure 5. Methods of detecting proteins using antibodies and of generating antibodies using proteins or synthetic peptides are known in the art and need not be described in detail herein. FasΔTM protein expression can also be monitored by measuring the level of mRNA encoding Fas-ΔTM. Any method for detecting specific mRNA species is suitable for use in this method. This is easily accomplished using the polymerase chain reaction (PCR) . Preferably, the primers chosen for PCR flank the TM region so as to provide a product that is measurably distinct in size between Fas and FasΔTM. Alternatively, Northern blots can be utilized to detect the specific mRNA species either by size or by probes specific to the mRNA encoding the TM region.
The invention also encompasses therapeutic methods and compositions involving treatment with FasΔTM. Either native or recombinant FasΔTM is suitable for use in this composition. Both should be substantially pure and free of pyrogens. It is preferred that the recombinant FasΔTM be produced in a mammalian cell line so as to ensure proper glycosylation. FasΔTM may also be produced in an insect cell line and will be glycosylated. For therapeutic compositions, a therapeutically effective amount of substantially pure FasΔTM is suspended in a physiologically accepted buffer including, but not limited to, saline and phosphate buffered saline (PBS) and administered to the patient. Preferably administration is intravenous. Other methods of administration include but are not limited to, subcutaneous, intraperitoneal, gastrointestinal and directly to a specific organ, such as intracardiac, for instance, to treat cell death related to myocardial infarction.
Suitable buffers and methods of administration are known in the art. The effective concentration of FasΔTM will need to be determined empirically and will depend on the type and severity of the disease, disease progression and health of the patient. Such determinations are within the skill of one in the art. Moreover, FasΔTM is a human protein normally found in the sera; administration of exogenous human FasΔTM is not likely to induce reactions such as anaphylactic shock or the production of antibodies. The upper concentration of FasΔTM for therapeutic use is thus not limited by these physiological considerations.
Administration of FasΔTM results in an increased extracellular concentration of FasΔTM which competitively binds the Fas ligand and therefore prevents or ameliorates apoptotic signals transmitted by Fas to the cell. The therapeutic method thus includes, but is not limited to, inhibiting Fas-mediated cell death. For instance, tumor necrosis factor (TNF) and Fas-specific antibodies are known to induce apoptosis and even whole animal death by binding to Fas. Inhibition of this interaction of Fas and ligands which induce it to trigger apoptosis thus will reduce apoptosis.
Suitable indications for therapeutic use of FasΔTM are those involving Fas-mediated cell death and include, but are not limited to, conditions in which there is inappropriate expression or up-regulation of Fas or the Fas ligand. Such indications include, but are not limited to, HIV infection, autoimmune diseases, cardiomyopathies, neuronal disorders, hepatitis and other liver diseases, osteoporosis, and shock syndromes, including, but not limited to, septicemia.
Methods of treatment with FasΔTM also include other mechanisms of increasing the extracellular concentration of FasΔTM. These include, but are not limited to, increasing cellular expression of FasΔTM. Suitable methods of increasing cellular expression of FasΔTM include, but are not limited to, increasing endogenous expression and transfecting the cells with vectors encoding FasΔTM. Cellular transfection is discussed above and is known in the art. Increasing endogenous expression of FasΔTM can be accomplished by exposing the cells to biological modifiers that directly or indirectly increase levels of FasΔTM either by increasing expression or differential processing of the FasΔTM over Fas or by decreasing FasΔTM degradation. Suitable biological modifiers can be determined by exposing cells expressing FasΔTM under the control of the native Fas promoter to potential biological modifiers and monitoring expression of FasΔTM. Expression of FasΔTM can be monitored as described above either by protein or mRNA levels. Biological modifiers can be any therapeutic agent or chemical known in the art. Preferably, suitable biological modifiers are those lacking substantial cytotoxicity and carcinogenicity.
Likewise, biological modifiers which reduce endogenous levels of FasΔTM are encompassed by the invention as is a method of increasing Fas-mediated cell death by decreasing endogenous levels of FasΔTM. The method of determining suitable biological modifiers is as discussed above, except that the endpoint is decreased levels of FasΔTM. Other methods of decreasing endogenous levels of FasΔTM include, but are not limited to, antisense nucleotide therapy and exposure to anti-FASΔTM antibody. Both these methods are known in the art and their application will be apparent to one of skill in the art. Suitable indications for decreasing endogenous levels of FasΔTM will be any which Fas-mediated cell death is appropriate. These include, but are not limited to, various types of malignancies and other disorders resulting in uncontrolled cell growth such as eczema. The following examples are provided to illustrate but not limit the present invention.
Example 1 Cloning of Fas-ΔTM Nucleic acid sequences encoding FasΔTM were cloned as follows. Unless otherwise specified, all cloning techniques were essentially as described by Sambrook et al. (1989) and all reagents were used according to the manufacturer 's instructions. mRNA was obtained from human lymphocytes and PCR was used to make cDNA specific for the FasΔTM mRNA. The lymphocytes were obtained and processed as follows. 35 ml of blood was obtained by venipuncture from a normal 42 year old male and immediately added to 350 μl 15% EDTA. 35 ml PBS was added to the blood and 17 ml of the blood suspension was layered on 12.5 ml Ficoll Paque (Pharmacia). This was then centrifuged at 1,800 rpm for 30 minutes in a swinging bucket rotor and a DynacII centrifuge. The plasma was aspirated and the lymphocyte layer collected and added to two volumes of PBS containing 0.9 mM Ca2+ and 0.5 mM Mg2+ (PBS/Ca/Mg buffer). The cells were washed once with the PBS/Ca/Mg buffer and resuspended in RPMI (Gibco/BRL) medium containing 2 g/1 glucose and 10% fetal bovine serum (FBS) at 2 x 106 cells/ml. The cell yield was 3.5 x 107 with greater than 98% viability. Concanavalin A (Sigma) was added to a final concentration of 30 μg/ml and the cells were incubated at 37°C for 72 hr. The cells were then processed for RNA isolation as follows. Total RNA was isolated from 3 x 106 cells using the guanidinium thiocyanate single-step RNA isolation method according to "Current Protocols in Molecular Biology" (1991) . The first strand cDNA used in the PCR reaction was synthesized from 6.4 μg of total RNA and resuspended in 100 μl water, according to the method described by Zapf et al. (1990) J. Biol. Chem. , 265:14892-14898. The FasΔTM cDNA was synthesized using PCR. The forward primer: 5'-GATTGCTTCTAGACCATGCTGGGCATCTGGACCCTCCTACC-3' contained an Xbal restriction site, and encoded the initiation methionine codon and first eight codons of Fas. The reverse primer:
5'-GTTGTTTGTCGACCTAGACCAAGCTTTGGATTTCATTTCTG-3' contained a Sail restriction site, and encoded the termination codon and last eight codons of Fas.
The PCR reaction was performed by adding to 1 μl of template cDNA (as described above, diluted 1:100), 100 pmoles of each primer, 2.5 units Amplitaq, 76.5 μl H20 and 10 μl buffer. The reaction proceeded at 94°C for 1 min, 55°C for 2 min, 72°C for 3 min for 35 cycles, 72°C for 7 min and was stored at 4°C. 15 μl of the reaction mix was loaded on a 1% agarose gel and a band of the appropriate molecular weight was detected. The remaining reaction mix was extracted with phenol/chloroform, ethanol precipitated and resuspended in 80 μl Tris-EDTA (TE) .
To the 80 μl sample the following was added: 5 μl Sail; 5 μl Xbal; 10 μl Sail reaction buffer. In another reaction, 60 μl H20 was added to 20 μl pBluescript at 0.65 mg/ml, 5 μl Sail. 5 μl Xbal and 10 μl Sail reaction buffer. Both tubes were incubated at 37°C for 2 hours and the reactions were run on a 1% preparative agarose gel. Bands corresponding to the digested DNA were excised from the gel and purified by Elutip® according to the manufacturer's instructions. The purified FasΔTM cDNA was resuspended in 20 μl TE buffer and the pBluescript in 40 μl TE buffer. The DNA samples were ligated in a reaction mixture containing 2 μl vector, 8 μl FasΔTM cDNA, 2 μl 10 mM ATP,
2 μl 10 x ligation buffer, 2 μl T4 DNA ligase (New England Biolabs) and 4 μl H20. The control reaction contained no FasΔTM DNA. After allowing the ligation to continue for 6 hr at 14°C the DNA was used to transform DH5α cells (Gibco) according to the manufacturer's instructions. Briefly, 200 μl cells were added to the ligation mix and kept on ice for 45 min. The cells were heat shocked for 90 sec at 42°C and then placed on ice.
3 ml L broth was then added and the cells were incubated for 1 hr at 37°C and plated on L broth agar plates containing 100 mM ampicillin, 20 μl 4% X-Gal and 50 μl 100 mM isopropyl-l-/3-D-thiogalactoside (IPTG) . The cells were allowed to form colonies by incubating overnight at 37°C. Positive colonies were grown overnight in L broth plus ampicillin, the plasmids were obtained by an alkaline lysis procedure and digested with Sail and Xbal according to the manufacturer's instructions. One plasmid containing the appropriate insert was prepared on a large-scale and the insert was sequenced by the dideoxy method. The sequence obtained is presented in Figure 3. The plasmid containing the recombinant cDNA encoding FasΔTM was designated pBluescript-FasΔTM.
Example 2
Analysis of Fas Genomic Structure The intron-exon organization in the Fas TM region was determined by PCR. Primers were designed to flank each of the putative introns, 1 and 2 (see Figure 2) . The forward and reverse primers flanking intron 1 were 5'-GATTGCTTCTAGAGGAATCA TCAAGGAATGCACACTC-3' and 5'-GTTGTTTGTCGACC CAAACAATTAGTGGAATTGGCAA-3' respectively, and the forward and reverse primers for intron 2 were 5'-AGATCTGCGGCCGCAT TGGGGTGGCTTTGTCTTCTTCTT-3' and 5'-GTTGTTTGTCGACGTTTTCCT TTCTGTGCTTTCTGCA -3' respectively. Xbal and Sail restriction enzyme sites and Notl and Sail restriction enzyme sites were included at the 5' ends of the intron 1 and 2 primers respectively to facilitate cloning of the PCR products. PCR was performed according to the manufacturer's instructions (Perkin Elmer Cetus) using human genomic DNA (Clontech) (5 μg) as template. 30 cycles of PCR were performed in a Perkin Elmer Cetus DNA Thermal Cycler with each cycle consisting of a 94°C, 1 min denaturation step, a 55°C, 2 min annealing step, and a 72°C, 3 min extension step. An additional 7 minute extension step was included after the last cycle. The PCR products were then incubated with 5 units of DNA poly erase I, Klenow fragment at 37°C, 30 min, extracted with phenol/chloroform/isoamylalcohol (1:1:0.04) followed by chloroform/isoamylalcohol (24:1) and recovered by ethanol precipitation.
The intron 1 and intron 2 PCR products were digested with Xbal and Sail and Notl and Sail respectively, agarose gel purified, ligated into pBluescript SK(-) and introduced into E. coli strain HB101 by the methods described in Example 1. Plasmid DNA was isolated using a Promega Magic miniprep kit according to the method described by the manufacturer. Plasmid DNA was sequenced directly by the dideoxy chain termination method using the Sequenase Version 2.0 DNA Sequencing kit according to the manufacturer's instructions (USB) . The sequence obtained is depicted in Figure 3.
Example 3 Expression of Recombinant Fas-ΔTM In order to express recombinant FasΔTM in the baculovirus system, the plasmid obtained in Example 1 was used to generate a second FasΔTM vector, designated pBlueBacIII-FasΔTM, by a PCR methodology as described in Example 1. The forward primer, 5'-TTTCCCGGATCCACAACCATGCTGGGCATCTGGACCCTCCTA-3' contained the convenient BamHI restriction site, and a Kozak consensus sequence ACAACC immediately preceding the initiation codon, and encoded the first seven amino acids of Fas. The reverse primer 5'-CCCCATGGCTAGACCAAGCTTTG GATTTCATT-3' encoded a termination codon, a Ncol site, and the last seven amino acids of Fas. Five recombinant plasmids were isolated. Two of them were sequenced by the dideoxy terminator method (Sanger et al. 1977) using sequencing kits according to the manufacturer's instructions (USB, Sequenase version 2.0). The DNA was sequenced using internal primers.
Clone 3, which did not contain any PCR errors in the sequence, was used to generate recombinant viruses by in vivo homologous recombination between the overlapping sequences pBlueBac III-FASΔTM-3 and AcNPV wild type baculovirus. After 48 hours post-transfection in insect Spodoptera frugiperda clone 9 (SF9) cells, the recombinant viruses were collected, identified by PCR and further purified. Standard protocols for plasmid cloning were employed (Maniatis et al. 1982) . Standard procedures for selection, screening and propagation of recombinant baculovirus were performed according to the manufacturer's instructions (Invitrogen) . After 48 hours post-transfection, the recombinant viruses were collected and purified. The molecular mass, on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-
PAGE) , of the protein produced in the baculovirus system is identical to the predicted molecular mass of FasΔTM according to the amino-acid sequence and the recombinant protein was also recognized by an anti-Fas antibody (Medical and Biological Laboratories, Nagoya, Japan) .
Example 4 Expression of FasΔTM in Mammalian Systems The FasΔTM coding sequence was excised from pBluescript-FasΔTM with Xbal and Sail, and introduced into plasmids pCEP7 and pREP7 (Invitrogen) at compatible Nhel and Xhol sites to generate clones FasΔTM-1 and FasΔTM-7, respectively. pCEP7 was generated by removing the RSV 3'-LTR of pREP7 with Xbal/Asp718. and substituting the CMV promoter from pCEP4 (Invitrogen) . To generate a compatible Xbal site, pCEP4 was first cleaved with Sail, and ligated to an oligonucleotide adapter containing an external Sail site, and an internal Nhel site. pCEP4 was then cleaved with Nhel and Asp.718 and the purified CMV promoter was ligated into pREP7 to generate pCEP7. 25 μg of each FasΔTM-containing plasmid was electroporated into the B lymphoblastoid cell line
WIL-2, and stable hygromycin resistant transformants were selected.
Example 5 Anti-Fas Antibody Induced Death of the Wild Type B Lymphoblastoid Cell Line WI-L2-729 HF2 and the Wild Type Cell Transformed by FasΔTM-1 and FasΔTM-7 2X105 WIL-2, FasΔTM-1 and FasΔTM-7 transformed WIL-2 cells were grown in RPMI supplemented with 10% fetal bovine serum (FBS) . After washing with fresh medium, the cells were suspended in RPMI supplemented with 10% FBS, 50 ng anti-Fas antibody was added, and the kinetics of cell death were analyzed by flow cytometry with FACScan. This method is based on the measurement of cells which shrink and are permeable to propidium iodide (PI) following their death. There was no difference in survival of all three cell lines in the control, but upon addition of anti-Fas antibodies, cells transformed by FasΔTM were less sensitive: by 26 hrs treatment approximately 50%, 40%, and 16% of wild type WIL-2,
FasΔTM-7, and FasΔTM-1 transformants died respectively (Fig. 5).
In another series of experiments, cells were initially grown for 24 hrs and then anti-Fas antibodies were added under the assumption that FasΔTM was secreted and should accumulate in medium. In this case the sensitivity of transformed cell lines was lower then without preincubation: after 26 hrs treatment with antibodies about 45%, 16%, and 5% of wild type WIL-2, FasΔTM-7, and FasΔTM-1 transformants died respectively (Fig. 6) .
To ensure that lower sensitivity of transformants is not caused by down regulation of Fas expression the amount of Fas on the surface of all three cell lines was compared. The cells were treated by standard procedures with monoclonal mouse anti-Fas antibodies, biotin-labeled anti-mouse IgM antibodies, and finally stained with FITC- labeled streptavidin. Analysis of stained cells using FACScan showed that there were no differences in the amount of Fas on the surface of wild type WIL-2 and the transformants (Fig. 7) . Thus WIL-2 transformed with FasΔTM are less sensitive to the cytotoxic effect of anti-Fas antibodies. This effect may be explained at least in part by secretion of FasΔTM by transformants. Thus, increased cellular expression of FasΔTM results in inhibition of Fas-mediated cell death.
Example 6 Recombinant FasΔTM Prevents Cell Death Induced by Anti-Fas Antibodies To check the biological activity of FasΔTM produced in a baculovirus system, insect cells transfected with wild type baculovirus and recombinant containing FasΔTM, as described in Example 3, were homogenized in water or in buffer containing 0.05% nonionic detergent Triton X-100 and centrifuged. Anti-Fas antibodies were preincubated 2 hrs at room temperature with aliquots of soluble and insoluble fractions, added to WIL-2, and cell death was analyzed after 24 hrs by flow cytometry as described above. There was no effect on cell viability of both soluble and insoluble fractions from insect cells transfected with the wild type baculovirus. At the same time soluble and insoluble fractions from insect cells transfected with baculovirus recombinant containing Fas-ΔTM inhibited death of WIL-2 induced by anti-Fas antibodies. Activity of the insoluble fraction was approximately ten times higher than that of the soluble fraction (Fig. 8) . Thus, recombinant FasΔTM can compete with Fas on the cell surface for binding antibodies and preventing Fas-mediated cell death.
Example 7 FasΔTM transcript analysis by RT-PCR Native FasΔTM transcripts were identified by RT-PCR and acrylamide gel electrophoresis. PCR primers were designed around the Fas TM region so that the Fas and FasΔTM transcripts would yield 296 bp and 233 bp PCR products, respectively. The forward primer was 5'-GACCCAGAATACCAAGTGCAGATGTA-3' and the reverse primer was 5'-CTGTTTCAGGATTTAAGGTTGGAGATT-3' . cDNA was synthesized from poly(A)+ or total RNA isolated from various human tissues and cell lines. These included heart, liver, activated and non-activated peripheral blood lymphocytes (PBLs) , placenta and fibroblast cell lines. PCR was performed as described in Example 1 using the cDNA as templates (10-100 ng/ml) and products were analyzed on 7% acrylamide/TBE gels.
All tissues and cell lines tested contained Fas and FasΔTM transcripts by this analysis. This suggests that cell death in these tissues can be modulated by the amounts (and ratios) of Fas and FasΔTM. Interestingly, liver contained the largest amounts of FasΔTM transcripts suggesting FasΔTM may be secreted into serum. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be apparent to those skilled in the art that certain changes and modifications may be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended claims.

Claims

We claim:
1. A composition comprising a purified nucleotide sequence encoding FasΔTM.
2. The nucleotide sequence according to claim 1 wherein the sequence is cDNA.
3. The nucleotide sequence according to claim 1, having the nucleotide sequence depicted in Figure 3.
4. A recombinant DNA vector comprising the nucleotide sequence according to claim 1.
5. The recombinant DNA vector according to claim 4 wherein expression of the sequence encoding FasΔTM is under control of an inducible promoter.
6. The recombinant DNA vector according to claim 4, selected from the group consisting of pBlueBACIII- FasΔTM-3, pBluescript-FasΔTM, FasΔTM-1 and FasΔTM-7.
7. A cell transfected with the recombinant DNA vector according to claim 4.
8. A transgenic animal comprising the recombinant DNA vector according to claim 4.
9. A substantially purified protein comprising the amino acid sequence of Figure 4.
10. A substantially purified protein comprising the Fas amino acid sequence but lacking a portion of the transmembrane region sufficient to result in a secreted protein that is not membrane bound.
11. The protein according to claim 10 wherein the protein is secreted.
12. The protein according to claim 10 wherein the protein is expressed by recombinant DNA.
13. The protein according to claim 10 wherein the protein is native protein.
14. An antibody which recognizes FasΔTM but not Fas.
15. The antibody according to claim 14 selected from the group consisting of polyclonal and monoclonal.
16. A composition comprising substantially purified FasΔTM and a physiologically acceptable buffer.
17. A method of detecting the presence of FasΔTM in a biological sample comprising the steps of a) obtaining the biological sample; b) adding anti-Fas antibodies to the biological sample; c) maintaining the biological sample under conditions that allow the anti-Fas antibodies to complex with FasΔTM; and d) « detecting the complexes formed.
18. The method according to claim 17 wherein the biological sample is sera.
19. The method according to claim 17 wherein the anti-Fas antibody is specific for FasΔTM.
20. A method for detecting the expression of DNA encoding FasΔTM in a biological sample comprising identifying the presence in the biological sample of mRNA encoding FasΔTM.
21. The method according to claim 20 wherein the method for identifying the FasΔTM mRNA is polymerase chain reaction and the primers flank the mRNA encoding the transmembrane region.
22. The method according to claim 20 wherein the method for identifying the FasΔTM mRNA is Northern blotting.
23. A method of treating Fas-mediated cell death in a patient comprising administering to a patient a therapeutically effective amount of FasΔTM.
24. A method of protecting cells from Fas-mediated cell death comprising the steps of increasing the endogenous concentration of FasΔTM.
25. The method according to claim 24 wherein the FasΔTM is exogenous.
26. The method according to claim 24 wherein the FasΔTM is expressed by the cell.
27. The method according to claim 26 wherein the FasΔTM is expressed by a recombinant gene.
28. The method according to claim 27 wherein expression of the gene is under the control of an inducible promoter.
PCT/US1994/013173 1993-11-15 1994-11-15 Novel fas protein and methods of use thereof WO1995013701A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP7514564A JPH09510864A (en) 1993-11-15 1994-11-15 Novel Fas protein and method of using the same
AU10569/95A AU1056995A (en) 1993-11-15 1994-11-15 Novel fas protein and methods of use thereof
EP95901253A EP0729300A4 (en) 1993-11-15 1994-11-15 Novel fas protein and methods of use thereof
KR1019960702558A KR960705929A (en) 1993-11-15 1994-11-15 Novel Fas Protein and Methods of Use (NOVEL FAS PROTEIN AND METHODS OF USE THEREOF)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/152,443 1993-11-15
US08/152,443 US5663070A (en) 1993-11-15 1993-11-15 Recombinant production of a soluble splice variant of the Fas (Apo-1) antigen, fas TM

Publications (1)

Publication Number Publication Date
WO1995013701A1 true WO1995013701A1 (en) 1995-05-26

Family

ID=22542947

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/013173 WO1995013701A1 (en) 1993-11-15 1994-11-15 Novel fas protein and methods of use thereof

Country Status (7)

Country Link
US (2) US5663070A (en)
EP (1) EP0729300A4 (en)
JP (1) JPH09510864A (en)
KR (1) KR960705929A (en)
AU (1) AU1056995A (en)
CA (1) CA2176575A1 (en)
WO (1) WO1995013701A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998013493A2 (en) * 1996-09-24 1998-04-02 Lxr Biotechnology, Inc. A family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
WO1999065935A2 (en) * 1998-06-18 1999-12-23 Karolinska Innovations Ab Fas peptides and antibodies for modulating apoptosis
US6015559A (en) * 1993-10-14 2000-01-18 Immunex Corporation Fas antagonists
US6235878B1 (en) 1996-07-19 2001-05-22 Takeda Chemical Industries, Ltd. Fas ligand-like protein, its production and use
US7576187B2 (en) 1996-09-24 2009-08-18 Genentech, Inc. Family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5888764A (en) * 1995-01-20 1999-03-30 Uab Research Foundation Human fas gene promoter region
AU4726797A (en) * 1996-10-31 1998-05-22 Mochida Pharmaceutical Co., Ltd. Prophylactic/remedial agent
US7285267B2 (en) 1997-01-14 2007-10-23 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6α & 6β
US20040013664A1 (en) * 1997-01-14 2004-01-22 Gentz Reiner L. Tumor necrosis factor receptors 6 alpha & 6 beta
US8003386B1 (en) 1997-01-14 2011-08-23 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6α and 6β
US7808479B1 (en) 2003-09-02 2010-10-05 Apple Inc. Ambidextrous mouse
WO1999040196A1 (en) * 1998-02-09 1999-08-12 Genentech, Inc. Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same
US20050037969A1 (en) * 1999-05-14 2005-02-17 Arbor Vita Corporation Molecular interactions in hematopoietic cells
US6942981B1 (en) * 1999-05-14 2005-09-13 Arbor Vita Corporation Method of determining interactions with PDZ-domain polypeptides
EP1270737A4 (en) * 2000-04-07 2004-06-16 Shionogi & Co Preincubation assay method
US7656393B2 (en) 2005-03-04 2010-02-02 Apple Inc. Electronic device having display and surrounding touch sensitive bezel for user interface and control
US11275405B2 (en) 2005-03-04 2022-03-15 Apple Inc. Multi-functional hand-held device
JP4578792B2 (en) * 2003-09-26 2010-11-10 富士通セミコンダクター株式会社 Solid-state imaging device
DK2406636T3 (en) 2008-12-10 2019-02-18 Joslin Diabetes Center Inc PROCEDURES FOR DIAGNOSTICATION AND PREVENTION OF RENAL DISEASE

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0510691B1 (en) * 1991-04-26 2004-11-03 Osaka Bioscience Institute DNA coding for human cell surface antigen
AU2515992A (en) * 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
WO1994008454A1 (en) * 1992-10-14 1994-04-28 Uab Research Foundation Methods, compositions and screening assays relating to autoimmune disease
IL111125A0 (en) * 1994-05-11 1994-12-29 Yeda Res & Dev Soluble oligomeric tnf/ngf super family ligand receptors and their use
JPH0875745A (en) * 1994-07-06 1996-03-22 Igaku Seibutsugaku Kenkyusho:Kk Immunoassay of soluble fas antigen and measuring kit therefor

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CELL, Volume 75, issued 17 December 1993, T. SUDA et al., "Molecular Cloning and Expression of the Fas Ligand, A Novel Member or the Tumor Necrosis Factor Family", pages 1169-1178. *
EUROPEAN JOURNAL OF IMMUNOLOGY, Volume 22, issued 1992, W.A.M. LOENEN et al., "The CD27 Membrane Receptor, A Lymphocyte-Specific Member of the Nerve Growth Factor Receptor Family, Gives Rise to a Soluble Form by Protein Processing that does Not Involve Receptor Endocytosis", pages 447-456. *
JOURNAL OF BIOLOGICAL CHEMISTRY, Volume 268, Number 22, issued 05 August 1993, K. HSU et al., "Differential Expression and Ligand Binding Properties of Tumor Necrosis Factor Receptor Chimeric Mutants", pages 16430-16436. *
KRAMER et al., "Apoptosis: The Molecular Basis of Cell Death", published 1991, by COLD SPRING HARBOR LABORATORY PRESS (N.Y.), pages 87-99. *
NATURE, Volume 364, issued 26 August 1993, J. OGASAWARA et al., "Lethal Effects of the Anti-FAS Antibody in Mice", pages 806-809. *
SCIENCE, Volume 261, issued 09 July 1993, N. ZHU et al., "Systemic Gene Expression After Intravenous DNA Delivery in Adult Mice", pages 20-211. *
See also references of EP0729300A4 *
THE JOURNAL OF EXPERIMENTAL MEDICINE, Volume 177, issued January 1993, E. ROUVIER et al., "Fas Involvement in Ca2+-Independent T Cell-Mediated Cytotoxicity", pages 195-200. *
THE JOURNAL OF EXPERIMENTAL MEDICINE, Volume 178, issued August 1993, J. WU et al., "Autoimmune Disease in Mice Due to Integration of an Endogenous Retrovirus in an Apoptosis Gene", pages 461-468. *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015559A (en) * 1993-10-14 2000-01-18 Immunex Corporation Fas antagonists
US6235878B1 (en) 1996-07-19 2001-05-22 Takeda Chemical Industries, Ltd. Fas ligand-like protein, its production and use
WO1998013493A2 (en) * 1996-09-24 1998-04-02 Lxr Biotechnology, Inc. A family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
WO1998013493A3 (en) * 1996-09-24 1998-06-04 Lxr Biotechnology Inc A family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US6433155B1 (en) 1996-09-24 2002-08-13 Tanox, Inc. Family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US7045604B2 (en) 1996-09-24 2006-05-16 Tanox, Inc. Family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US7045596B2 (en) 1996-09-24 2006-05-16 Tanox, Inc. Family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US7576187B2 (en) 1996-09-24 2009-08-18 Genentech, Inc. Family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
WO1999065935A2 (en) * 1998-06-18 1999-12-23 Karolinska Innovations Ab Fas peptides and antibodies for modulating apoptosis
WO1999065935A3 (en) * 1998-06-18 2003-04-17 Karolinska Innovations Ab Fas peptides and antibodies for modulating apoptosis
US6846637B1 (en) 1998-06-18 2005-01-25 Imed Ab Fas peptides and antibodies for modulating apoptosis

Also Published As

Publication number Publication date
US5663070A (en) 1997-09-02
AU1056995A (en) 1995-06-06
EP0729300A1 (en) 1996-09-04
US5652210A (en) 1997-07-29
JPH09510864A (en) 1997-11-04
KR960705929A (en) 1996-11-08
EP0729300A4 (en) 1998-08-05
CA2176575A1 (en) 1995-05-26

Similar Documents

Publication Publication Date Title
US5652210A (en) Soluble splice variant of the Fas (APO-1) antigen, FasΔTM
JP4793836B2 (en) Fibroblast growth factor-like polypeptide
CA2343655C (en) Interleukin 17-like receptor protein
KR100391227B1 (en) Dna
BG65519B1 (en) Interleukin-18-binding proteins, methods for their preparation and administration
CZ29397A3 (en) Chemokins originating from macrophages and their analogs, polynucleotides encoding thereof, use of such chemokins and pharmaceutical compositions based thereon
BG64779B1 (en) Ligand related to tumour necrotic factor
JP4326944B2 (en) Surface molecule NTB-A involved in natural killer cell activity
JPH11225774A (en) Member of immunoglobulin gene superfamily, pigr-1
AU2002325333A1 (en) NTB-A, a surface molecule involved in natural killer cells activity
US6090575A (en) Polynucleotides encoding human G-protein coupled receptor GPR1
WO1993000357A1 (en) Therapeutic polypeptides based on von willebrand factor
EP0524173B1 (en) Solubilization and purification of the gastrin releasing peptide receptor
WO1998033819A1 (en) Cellular receptors for subgroup c adenoviruses and group b coxsackieviruses
US6030804A (en) Polynucleotides encoding G-protein parathyroid hormone receptor HLTDG74 polypeptides
WO1998033819A9 (en) Cellular receptors for subgroup c adenoviruses and group b coxsackieviruses
AU1793092A (en) Receptors for bombesin-like peptides
JP2001504336A (en) Connective tissue growth factor-3
EP0648268A1 (en) Therapeutic fragments of von willebrand factor
US6130061A (en) Human stem cell antigen 2
WO1995004756A1 (en) Complement inhibitor proteins of non-human primates
US20020049304A1 (en) Human CCN-like growth factor
US8110659B1 (en) Human tumor necrosis factor receptor-like genes
JP2003520590A (en) Chondromodulin I-related peptide
CA2206640A1 (en) Human vascular ibp-like growth factor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW NL NO NZ PL PT RO RU SD SE SI SK TJ TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2176575

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995901253

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1995901253

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995901253

Country of ref document: EP